You are on page 1of 11

Miquel et al.

Microbial Cell Factories (2015) 14:48


DOI 10.1186/s12934-015-0229-1

REVIEW Open Access

A proposed framework for an appropriate


evaluation scheme for microorganisms as novel
foods with a health claim in Europe
Sylvie Miquel1,2, Martin Beaumont1,2, Rebeca Martín1,2, Philippe Langella1,2, Véronique Braesco3 and Muriel Thomas1,2*

Abstract
This paper concerns the procedure and the scientific approach to obtain market authorization for a microorganism
to be recognized as a novel food with a health claim. Microorganisms that have not been traditionally used during
food production in Europe prior to 1997 are considered as novel foods, which should undergo an in-depth
characterization and safety assessment before being authorized on the European market. If a novel food bacterium
is claimed to provide a beneficial effect on health, these claims must also be investigated before they can be authorized.
Some requirements to obtain novel food certification are shared with those required to obtain a health claim. Although
regulation exists that deals with these issues for foods in general, bacteria in food raise a specific set of questions that
are only minimally addressed in official documentation. We propose a framework and suggest a list of criteria that
should be assessed to obtain marketing authorization and health claim for a bacterium in accordance with European
health policy.
Keywords: Beneficial microbes, Probiotic, Regulation, Safety, Gastro Intestinal (GI) tract

Introduction in theory be considered in itself as a health claim [3]. For


The introduction of high throughput sequencing, ad- instance, the Food Safety Authority of Ireland clearly indi-
vanced bioinformatics, and specialized in vitro and cates on its website that the “term probiotic is considered
in vivo models has improved the understanding of to be a health claim”.
mechanisms underlying the action of probiotics. Probio- Emerging clinical evidence suggest that beneficial bac-
tics are defined as “live microorganisms which when ad- teria positively influence a wide range of human health
ministered in adequate amounts, confer a health benefit issues, especially digestive health [4-7]. Today, most mi-
on the host” [1]. For grammatical reasons, this definition croorganisms marketed as “probiotics” or beneficial bac-
has been recently re-worded by an expert panel of the teria by the food industry belong to the genera
International Scientific Association for Probiotics and Lactobacillus and Bifidobacterium [8]. However, these
Prebiotics (ISAPP) as, “live microorganisms that, when genera are sub-dominant in the intestinal microbiota in
administrated in adequate amounts, confer a health bene- adults. This observation, in association with rapidly
fit on the host” [2]. Since February 2013, the European expanding knowledge of the human microbiome, sug-
Commission no longer allows companies to communicate gests that a large panel of potential new candidates can
assumed health benefits of products based solely on pro- be isolated from the dominant members of our adult
biotic content. Yet, the use of the term “probiotic”, which microbiota. The real challenge for translational projects
is not regulated in Europe, implies that the product has a between scientists and industrial partners will be the
beneficial health effect, and thus this designation should introduction of new generations of beneficial strains be-
cause there is currently a large gap between the bench
and the market. It is indeed difficult for all stakeholders,
* Correspondence: muriel.thomas@jouy.inra.fr
1
Commensal and Probiotics-Host Interactions Laboratory, UMR1319 Micalis,
including academic and industrial partners, to agree
INRA, AgroParisTech, Domaine de Vilvert, 78350 Jouy en Josas, France unanimously on a system of regulation, which may need
2
AgroParisTech, UMR 1319 MICALIS, F-78350 Jouy-en-Josas, France to be adjusted on a case-by-case basis.
Full list of author information is available at the end of the article

© 2015 Miquel et al.; licensee BioMed Central. This is an Open Access article distributed under the terms of the Creative
Commons Attribution License (http://creativecommons.org/licenses/by/4.0) which permits unrestricted use, distribution, and
reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain
Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article,
unless otherwise stated.
Miquel et al. Microbial Cell Factories (2015) 14:48 Page 2 of 11

This review focuses on foods and food ingredients Table 1 Statutory text, official documents of the EFSA
consisting of or including live bacteria and does not Novel Food
cover genetically modified microorganisms, which raise Recommendation Related to the scientific aspects and the
other issues. Bacteria with no history of documented safe 97/618/EC presentation of information necessary to support
use in Europe prior to 1997 are classified as novel foods. applications to place novel foods and novel food
ingredients on the market
Requirements that must be fulfilled for such bacteria
Regulation Related to novel foods and novel food ingredients of
to be allowed on the market include the accurate N° 258/97 the European Parliament and of the European Council
characterization of the strain and a solid demonstra-
Novel Food Complex Novel Food from non-Genetically Modified
tion of its safety. If a novel food confers a beneficial ef- Class 2 sources according to Categories of novel foods and
fect on health, it will become a novel food associated novel food ingredients identified in Regulation (EC)
with a health claim after approval by the EFSA and No 258/97. Intact plants, animals and microorganisms
used as foods as well as food components (e.g.
authorization from the European Commission. How- complex carbohydrates, fats, proteins or those
ever, it remains difficult to define experiments that can substances collectively described as dietary fiber)
be judged as reliable, valuable, and pertinent to sup- are included. Two sub-classes can be identified:
port the application of novel bacteria. Here, we 2.1 the source of the NF has a history of use in
summarize criteria that are commonly suggested by food in the Community.
authorities or in the scientific literature for the 2.2 the source of the NF has no history of use in
food in the Community.
characterization of novel bacteria and the assessment
of their safety and efficacy. We also present our point Health claim
of view about the biological relevance and regulatory Regulation Related to nutrition and health claims and establishes
N° 1924/2006 rules governing the Community authorization of
significance of these criteria and of the experimental health claims should only be authorized in the
methods often proposed to meet them. Community after a scientific assessment of the highest
possible standard, to be carried out by the EFSA.
What is a novel food according to the current European
food regulation?
Many microorganisms that are used as food ingredients sample), as a novel food supplement in the European
or food additives have a long history of safe use in food Union (EU) (Miya-Pro; Public version – Clostridium
fermentations [8]. However, microorganisms that were butyricum 588 novel food application).
not traditionally used in food production in Europe be-
fore 1997 are classified as a novel food class 2.2 (see
Table 1 and Figure 1). The market and the legislation of What is a novel food with a health claim according to the
novel foods are regulated by the UE 97/618/EC recommen- current European food regulation?
dation and regulation No 258/97 (Table 1 and Figure 1). A novel food is not required to have any beneficial nutri-
The launch a Novel Food in Europe is authorized if the tional or health-related effects upon consumption. How-
product has been thoroughly characterized and its safety ever, a health claim would highlight the benefits of the
has been proven (Figure 1). novel food for the consumer and may help its commer-
For a novel food to be allowed on the market, those cial success (Figure 2). The process for substantiating a
submitting the application are required to do the health claim for a novel food is the same as for a con-
following: ventional food. Beneficial nutritional or health claims
may be communicated to the consumer only after
# Characterize the microorganism and its metabolites authorization from the European Commission, which re-
# Characterize how the process of production may quires a favorable opinion from the EFSA according to
modify the microorganism EU regulation 1924/2006 (see Table 1 and Figure 1). As
# Give the history of production and consumption of shown in Figure 2, some requirements to obtain novel
the microorganism food certification are the same as those required to cer-
# Anticipate the intake/extent of the use and the tify a health claim (e.g. strain characterization).
consumption of the novel food Overall, most bacteria that will be used in foods for
# Provide the nutritional composition of the novel food human consumption in the near future will need to
# Assess the safety of the microorganism comply with two different regulations (EC 258/97 and
# Give toxicological information EC 1924/2006), which largely involve scientific require-
ments. Successful market applications will require vari-
An example of an application for a novel food is the ous skills allying the academic and industrial worlds, to
public version of an application for the use of Clostridium address the numerous regulatory, economic, and scien-
butyricum (CBM588), (which was isolated from a soil tific challenges.
Miquel et al. Microbial Cell Factories (2015) 14:48 Page 3 of 11

For future foods and food ingredients consisting of or isolated from bacteria

Could you prove the


consumption within the Yes No
EU before 15 May 1997

Is it a Novel Food ? No Yes

Do you need to fulfil the UE


No Yes
258/97 ?

For future health claims

Do you need to fulfil


Yes Yes
The UE 1924/2006?
Figure 1 Statutory text.

How can current regulation be improved to ensure the that it may encounter from the start of the production
correct characterization of future novel-food microorganisms process until the final consumption of the product.
with a health claim? The EFSA recommends that both the species (DNA-
Characterization of the strain of microorganism is the DNA hybridization or 16S rRNA gene sequence analysis)
first requirement both to apply for novel food market- and strain (genetic typing molecular methods) be identi-
ing authorization and to submit a health claim request fied [12]. Indeed, it is essential to identify clearly the spe-
(Figure 2). Functional effects are only related to the cies and strain of bacteria based on phenotypic and
particular strain and cannot be extended to the rest of genotypic data [13] from various methods considered as
the species to which the strain belongs [9,10]. However, reliable references in the literature (Table 2) [14]. Al-
the only approved health claim is for yoghurt cultures, though all methods listed in Table 2 are relevant, some
which are not defined at the strain level [11]. Thus, if may be replaced by whole genome sequencing technolo-
there is a reasonable scientific basis to justify a health gies, which are undergoing rapid development. In
benefit for an entire species, then the health claim need addition to characterizing the strain, the sequence may
not be limited to one strain. Moreover, the ISAPP re- reveal the pathogenic potential of the strain and identify
cently proposed the creation of a general category of virulence genes; thus, this information may be relevant
probiotics defined at the species level and associated for the toxicological assessment required for a novel
with core benefits [2]. It is also important to consider food. Additionally, genomic data are useful to search for
characterizing the strain in the various environments functional relationships between genetic elements and
the mechanisms underlying probiotic action [15]. Puta-
tive proteins and metabolic pathways can also be pre-
Novel foods Health claims
dicted from genomic sequences and these predictions
can be confirmed by metabolomic and transcriptomic
EU 258/97 EU 1924/2006
profiling with high-throughput techniques. In light of
Compulsory Optional the development of many “omic” techniques, it would be
interesting to establish a strain-specific map of genomic
and functional interactions. In the near future, it is prob-
able that most bacteria that will be used for the first time
Consumption in foods for human consumption will have to be entirely
history Health effect
Characterization evidence
sequenced.
Safety Clearly, whole genome sequencing is the gold standard
for characterization; however, we can only make sense of
these data if we consider a strain in its physiological
context and take into account its overall biology. Life
cycle, industrial processes, storage history and ingestion
may affect the phenotypic properties of bacteria and
Bringing to market Sales success
the potential activities of a particular strain [16]. It
could be worthwhile to describe the properties of
Figure 2 Novel food or health claim?
microorganisms during different physiological states
Miquel et al. Microbial Cell Factories (2015) 14:48 Page 4 of 11

Table 2 Criteria and methods often proposed to characterize a strain (required for both novel food and health claim
regulations)
Taxonomic Biological Methods Propositions for improvement
level assignment
Species Phenotypic GRAM staining
Morphologic description The physiology of the bacterium in environments
from production to consumption should be
extensively characterized
Analytical profile identification (API)
Genotypic 16S rRNA sequencing [15,33] Full genome sequencing should be systematically
provided
MLST (Multi Locus Sequence Typing)
Strain Metabolic Analytical profile identification (API)
Genotypic Genome sequencing [15]: Identification of virulence genes
Pulsed field gel electrophoresis (PFGE) [20]: evaluation of
genetic stability [58]

(latency, multiplication, and stationary phases) but also Concerning intake, it is strongly recommended that
in the context of the various micro-environments that the final products contain an adequate amount of live
the product could encounter. However, the exact na- bacteria to provide a health benefit [24,25]. A daily in-
ture of such tests cannot be generalized and specific take of at least 108–109 viable cells, which may be
tests are needed for each microorganism used. For in- achieved by the daily consumption of at least 100 g of
stance, the yoghurt bacteria, Streptococcus thermophilus final product, has been suggested as the minimum intake
and Lactobacillus delbrueckii ssp. bulgaricus present to provide an effect [24].
different growth rates and metabolic activities depend-
ing on the culture media (milk, presence of lactose or How should the safety of microorganisms and their
rich media) [17-19]. Although the environment may in- metabolites be assessed? Some suggestions to improve
fluence the metabolic activities and/or functions of mi- current regulatory requirements
croorganisms the inverse is also true: microorganisms may In the EU, the a priori safety of some microorganisms is
affect the physico-chemical conditions of their environ- accepted if they benefit from Qualified Presumption of
ment. Moreover, strains could evolve and functional muta- Safety (QPS) status [26].
tions could appear. Thus, pulsed field gel electrophoresis Most of these microorganisms are Gram positive, non-
(PFGE) may be informative because a high rate of muta- sporulating or lactic acid bacteria [24]. Lactic acid bacteria,
tions may lead to the appearance of new characteristics mainly Bifidobacterium, Lactobacillus and Streptococcus
[20]. This is mainly a typing technology, yielding limited are found in many food products and are not dominant in
data on functionality. Most (functionally-important) SNPs the intestinal microbiota in adults [8,22,27-29]. If the
are actually not detected by rare cutting restriction en- microorganism is not recognized as QPS, a complete as-
zyme analysis. Thus, novel-food bacteria should also be sessment of its safety must be carried out according to
highly characterized in the various environments that they regulatory requirements. Some adverse side effects that
will encounter from production to consumption. must be monitored are the production of host-deleterious
The final product of the food matrix in which the metabolites, systemic infection, inappropriate immune re-
microorganism is present should also be precisely sponses, antibiotic (AB) resistance and gene transfer [30].
characterized: However, this list is not exhaustive and there is currently
no official document summarizing these criteria. Some re-
– to assure the absence of microbial contaminants [21]; quirements are mentioned in the scientific literature
– to define the composition of the food because it may [21,31,32] and in the Guidelines for the Evaluation of Pro-
affect strain growth; biotics in Food published in 2002 by the FAO/WHO
– to assure proper labeling of the food with regard to working group [13]. However, the biological relevance of
macro-nutrients, calories and allergens, especially these requirements remains a subject of debate. We have
for particular consumers (e.g. diabetics) [22]; ordered and evaluated the criteria that are often proposed
– to define clearly the conditions of use (the target to demonstrate the safety of microorganisms into five clas-
population, the storage conditions and any ses: survival and/or viability along the Gastro Intestinal
precautions about the time and regularity of intake) (GI) tract, effect on intestinal homeostasis, adhesion,
and to support its claimed effect [23]. metabolic activities, and remote effects (Table 3).
Miquel et al. Microbial Cell Factories (2015) 14:48
Table 3 Common criteria generally considered as essential for the safety of NF/probiotic products (required for both novel food and health claim regulations)
What How Why Comments and Propositions for improvement
Survival in GI tract Resistance to intestinal stress In vitro Growth curves/Detection in feces Resistance to GI tract conditions may Not valuable for all beneficial effects
conditions after consumption favor the beneficial effects
Development of new protectors/encapsulators
Bile salt deconjugation High-performance liquid chromatography Large amounts of deconjugated bile Evaluation of property in vitro has poor relevance;
salts may have undesirable effects assessment of bile salt deconjugation in vivo
on the human host
Mass spectrometry
Preservation of the Microbiota Perturbation of commensal In vitro production of bacteriocins or Bacteriocins and AB may perturb Development of growth inhibitory references
homeostasis of gut consortium antibiotics (AB) microbiota. with major commensal bacteria
barrier components
AB may interact with a patient’s
treatment
Antibiotics (AB) resistance In silico*prediction and in vitro antibiogram AB resistance may be transmitted Development of in vivo assessment (animal model)
between bacteria indicating the microbiota homeostasis (composition
Minimal inhibitory concentration test (MIC) and activities) after probiotic consumption
Minimal bactericidal concentration test If plasmids are detected: the presence/absence of
(CMB) genes encoding the most common resistance
determinants should be characterized
Presence of plasmids In silico*prediction or DNA extraction Plasmids favor the transmission of Requirement to up-date the antibiotic list
followed by analysis by gel electrophoresis antibiotic resistance
Mucus Mucus degradation Mucin degradation test (agarose gel or Excessive mucus degradation may The capacity to degrade mucus seems to be a poor
liquid culture) lead to intestinal barrier weakening criterion to estimate the protective or deleterious
effect of bacteria on the intestinal barrier
Adhesion and Intestinal/Mucosal adhesion Test bacterial strain adhesion to epithelial Mucosal adhesion may interfere with The intestinal/mucosal adhesion capacity can be
translocation risk cell line pathogenic microorganisms, stimulate either a beneficial or a deleterious criterion
beneficial cellular processes, or favor
bacterial translocation
Intestinal mucosa degradation Gelatinase activity assay Mucosal degradation may weaken It could be useful to evaluate in vivo
the intestinal barrier translocation capacities
Hemolytic activity Blood agar culture Hemolysis damages red blood cells
Metabolic activities D-Lactate production Colorimetric assay D-Lactate accumulation in blood The production of D-lactate should be compared
leads to acidosis with the amount produced by usual strains
(like in yoghurt)
Toxin production Protocol recommended by the European Toxic molecules Establishment of threshold values relevant in
scientific committee of animal nutrition. humans
Biogenic amine production Colorimetric assay Immune responses such as allergic
responses
Remote effects Platelet aggregation Aggregation test Risk of thrombosis Development of ex vivo protocols (explants,
organoids)
Genotoxicity Toxicity testing on animals models (chronic Risk of cancer

Page 5 of 11
and subchronic tests)
Allergenicity Allergic response
(*: if the complete genome is available).
Miquel et al. Microbial Cell Factories (2015) 14:48 Page 6 of 11

Survival and /or viability in the intestinal tract strains inhibit the growth of commensal bacteria, to as-
It is commonly accepted that bacteria must be able to sess whether and how homeostasis is maintained. This
survive in at least the upper part of the GI tract to have approach requires the assessment in vitro of the produc-
a beneficial effect on the host (except bacteria used to tion of bacteriocins or antibiotics (AB). It would be use-
improve microbial composition in the oral cavity). An ful to test in vitro if probiotics have bactericidal effects
intestinal strain would have to resist extreme GI tract on the main groups of commensal bacteria such as Bac-
conditions: an intestinal pH gradient (from 4.0 in stom- teroidetes and Clostridii, although this is not required by
ach to 7.0 in the lower part of intestine), bile salts, and authorities and is rarely proposed in the literature. Then,
pancreatic secretions [33]. All these variables can be the effect of the probiotic consumption on microbiota
monitored in vitro by growth curves, although such as- composition could be assessed with in vivo experiments
says do not totally reflect in vivo conditions (Table 3). measuring the global metabolic activities of microbiota
The amount of bacteria in vivo can also be determined (such as the production short-chain fatty acids).
by PCR or by isolating and culturing them from stools a Novel food strains should not be able to transmit anti-
few days after ingestion [34]. However, it is debatable biotic resistance genes to bacteria in their environment
whether a bacterium must survive and proliferate along to avoid the acquisition and spread of multiple antibiotic
the GI tract to have an effect. For example, polysacchar- resistance [33]. The transmission potential of resistance
ide A of Bacteroides fragilis protects animals from colitis genes depends on their genetic support (plasmids or
induced by Helicobacter hepaticus [35] and the peptido- chromosome). Unlike resistance genes carried by plas-
glycan of the Lactobacillus salivarius strain Ls33 also mids, those carried by chromosomes have a much lower
protects against experimentally-induced colitis [36]. risk of transfer. Thus, it is important to consider the
Thus, proliferation of the bacterium at the target site is genomic location of an antibiotic resistance gene when
not absolutely required for its effect and a daily con- testing a strain for antibiotic resistance [32]. A list of an-
sumption of product may suffice. Nonetheless, survival tibiotics which should be tested to assess antibiotic re-
in the GI tract needs to be examined in each particular sistance has been proposed by the EFSA [41]. This list
case, and it would be informative to establish a sensitiv- should be updated according to current treatments and
ity profile of each strain towards pH, bile salts, and pan- the target population (Table 3).
creatic secretions. Innovative technologies, based for It is also widely reported that bacteria are beneficial if
example on encapsulation, are currently being developed they do not degrade mucus. The rationale is that degrad-
to control better the viability of microbes during storage, ing mucus weakens the intestinal barrier and conse-
processing, and in the GI tract [37]. quently destabilizes the protective function of the
The deconjugation of bile salts is a property that al- epithelium [42]. However, some commensal bacteria use
lows bacteria to survive in the GI tract and is implicated mucins, the major constituents of mucus, as an energy
in lowering circulating cholesterol levels [38]; however, source and can stimulate host mucus production [43].
in excess, this process may be deleterious for the host Thus, such adaptive cross-talk does not necessarily im-
[39]. Thus, it has been proposed that the bile salt decon- pair host defense mechanisms. Moreover, mucin degrad-
jugating activity of probiotics should be evaluated [40], ation tests (agarose gel or liquid culture) often use
because it could influence bacterial survival and host synthetic mucins from pig gastric mucus. This experi-
health. Evaluation of this property (Table 3) in vitro may mental setting does not accurately represent the typical
provide little relevant information for the situation features of human intestinal mucus, and moreover, the
in vivo because of its complexity, raising the need, as a diversity of available energetic substrates in vivo is much
second step, to perform experiments with an animal higher than in vitro. Thus, bacteria that degrade mucus
model (mono-associated or with a complete microbiota). in vitro (where mucus is the only energy source in cul-
It is yet not completely clear whether bile salt deconju- ture medium) may not necessarily use this substrate
gating activity is a desirable trait in a novel food/pro- in vivo. In our opinion, the capacity to degrade mucus,
biotic bacterium. In our opinion, the capacity to as assessed by in vitro assays, is not a relevant criterion
deconjugate bile salts is informative when choosing a to estimate the protective or deleterious effect of bac-
strain and should therefore be considered. teria on the intestinal barrier (Table 3). For instance, ad-
ministration of living Akkermansia muciniphila, which is
Preservation of intestinal homeostasis by maintaining the a mucin-degrading commensal, reverses dietary induced
integrity of two barrier components of the gut: microbiota metabolic disorders [44].
and mucus
A key safety requirement is that the use of a new novel Adhesion properties and translocation risk
food should not perturb the population commensal or- Adhesion to the mucosal layer is commonly mentioned
ganisms. It may be useful to examine whether novel food as a factor favoring durable implantation and a highly
Miquel et al. Microbial Cell Factories (2015) 14:48 Page 7 of 11

effective probiotic [13]. This suggests that transient per- It remains informative to determine the level of D-
sistence and/or long-term colonization is associated with lactate production for each microorganism, but the
beneficial mechanisms such as interfering with the production of D-lactate should not be considered sys-
growth of pathogenic or potentially pathogenic microor- tematically as a metabolic disadvantage for future
ganisms in the body or stimulating other potentially health-related strains. It could be interesting to com-
beneficial cellular processes [45]. In our opinion, this as- pare the amount of D-lactate produced by the consid-
sumption is questionable, because proximity to the in- ered strains with that produced by Lactobacilli currently
testinal mucosa and a long transit time in the gut are present in yoghurt.
not sufficient to maximize the beneficial effects of a Lack of toxin or biogenic amine production is essential
strain (Table 3). Indeed, although bacteria are rapidly to ensure strain safety but experimental settings and
eliminated, they may act transitorily [46]. For instance, threshold values are still poorly defined for human appli-
the immune system of germ-free mice can be stimulated cations. The EFSA recommends a protocol originally
by temporary bacterial colonization [47]. These results established for animal nutrition and limited to the use of
imply that some microorganisms do not have to colonize Bacillus species [54]. This may be too specific to be ex-
permanently the microbiota to affect host responses trapolated to all strains used in human nutrition or
[46]. In addition, in vitro tests have many limits, because health. A relevant alternative strategy may be the use of
adhesion properties and mucus production depend on commensal bacteria as references (Table 3).
the cell line under study [48]. We believe that although Two other tests are frequently mentioned: strain
adhesion is an important characteristic of a strain, it hemolytic activity and platelet aggregation ability. Both
should not be a criterion to estimate the potential effect are typical features of pathogenic bacteria (Table 3).
within the gut. However, in vitro tests that assess adhe- However, these deleterious effects may only happen if
sion also reveal the cytotoxicity of the bacterium on tar- the ingested bacteria end up in the blood. This is an un-
geted cells. It may be relevant to estimate bacterial likely situation which requires bacterial translocation
translocation because oral treatments containing a high across a weakened intestinal barrier, which would also
dose of probiotic may be deleterious, especially in im- favor the translocation of all commensal bacteria. These
munodeficient patients [49]. tests however provide important information about
strain pathogenicity [55-57].
Metabolic activities: threshold and physiological state matter
It is well known that some bacterial metabolites have Remote effects: toxicology testing
deleterious effects; however, the current recommenda- It is necessary to evaluate the toxicological profile, in-
tions provided by authorities are unclear and no max- cluding genotoxicity, to establish the safety of a new
imal threshold for these metabolites has been proposed product [58]. The harmful effects of a particular sub-
(Table 3). For instance, bacteria-induced D-lactate pro- stance or microorganism are examined, evaluated, and
duction can be viewed as harmful because the accumula- interpreted by testing it on animal models (Table 3).
tion of this metabolite in blood may be neurotoxic and These results are then extrapolated to determine the
may lead to acidosis [50]. Individuals the most at risk quantity that will produce similar effects in humans.
are those with short-bowel syndrome (which results However, current ethics statements encourage a reduc-
from resection of the small intestine) because of the ac- tion in the numbers of animals that are used in these
cumulation D-lactate in their feces [51]. In healthy tests. Thus, it may be useful to develop new techniques
adults, lactate is not detectable in fecal samples because based on viable ex vivo tissue explants or organoids
lactobacilli (main producers of D-lactate) represent a (from experimental rodents or humans) that could be
minor group in the microbiota and lactate is degraded co-incubated with bacteria of interest or their respective
by other major bacterial groups [52]. Some strains of L. metabolites [59,60]. In our opinion, ex vivo intestinal
delbrueckii subsp. bulgaricus that are widely used in yog- cultures co-incubated with bacteria or their culture
hurt fermentation produce D-Lactate [17]. The subse- supernatant are promising approaches that should be
quent risk of acidosis related to D-lactate after the encouraged to evaluate immune responses (cytokine re-
ingestion of yoghurt is not a health threat in healthy in- lease in the supernatants and the expression level of
dividuals [53]. Nearly all lactic acid bacteria, even those immune receptors), epithelium homeostasis (prolifera-
widely used for food applications, produce the D-lactate tion, differentiation and/or apoptosis) and toxicity
isoform. Moreover, many commensal strains present in markers (e.g. genotoxicity) of new products.
the lower GI tract consume D-lactate resulting in cross- Overall, although in vitro testing can be very inform-
feeding that may help to explain the reported butyro- ative as a first step, the properties of candidate probio-
genic effect of certain dietary substrates [52]. Thus, the tics should always also be assessed in vivo because they
production of D-lactate is not a risk for healthy people. are strongly influenced by the intestinal ecosystem, the
Miquel et al. Microbial Cell Factories (2015) 14:48 Page 8 of 11

complexity of which cannot be entirely reproduced The intestinal microbiota: a potential source of novel
in vitro (biochemical properties, interaction with com- foods with a health claim
mensal microorganisms or with the host and substrate The intestinal microbiota, including commensal bacteria
availability). and probiotics, plays a fundamental role in the develop-
ment and maintenance of intestinal homeostasis by par-
ticipating in the immune and digestive functions of the
Are health claims based on a cause-effect relationship GI tract [33]. This homeostasis is crucial for the host
between the intake of a microorganism and a benefit to health and if disrupted, it may lead to an inappropriate
human health? reaction of the immune and digestive systems.
Clearly, the safety of a microorganism is essential for the Newly discovered intestinal bacteria may be used for
authorization of a novel food. However, convincing evi- the development of new novel foods containing microor-
dence demonstrating the beneficial effects of a particular ganisms with a health claim [2]. Over the past 20 years, in-
microorganism should also be submitted for scientific tensive research has led to the in depth characterization of
evaluation to the Nutrition panel of the EFSA. Numer- intestinal communities, particularly as a function of age,
ous general and specific guidelines (depending on the health status, geographic location, nutritional habits, med-
health claim) have been published by the EFSA and ical care, and genetic predisposition of the host [64]. It is
should be consulted and followed [23,61-63]. Health ef- now widely accepted that intestinal commensal microor-
fects are demonstrated in the same way regardless of ganisms participate in the physiology and the health of
the nature of the active component of the novel food their host through metabolic, protective, and trophic func-
(microorganism, nutrient, or any other ingredient). tions [65]. Host physiology, gut maturation, innate and ac-
Successful applications preferably involve studies of quired immune responses and metabolism are largely
high methodological and statistical quality examining a influenced by the metabolic properties of the microbiota
well-defined health benefit that can be unambiguously [66-69]. The activity and composition of the microbiota
evaluated through changes in a recognized biomarker are modulated by external factors, such as diet, making it
in humans. a highly “malleable” tissue in humans [70]. It was recently
It is particularly difficult to define the beneficial effects proposed that Faecalibacterium prausnitzii, which is a
after consumption of a bacterium. The ISAPP considers major constituent of the intestinal human microbiota,
that beneficial microbes that support a healthy digestive may have prophylactic or therapeutic applications in
tract and a healthy immune system are associated with human health [71,72]. In particular, F. prausnitzii is de-
common general benefits to human health [2]. Indeed, pleted in many intestinal disorders and displays benefi-
many of the most promising benefits of bacteria involve cial anti-inflammatory effects on host, suggesting that
their effect on the gut, and their interactions with either it may be used to counterbalance the dysbiosis linked
the gut microbiota or the cells of the intestinal mucosa, to certain diseases [73,74]. Thus, the characterization
especially epithelial and immune cells. These aspects of several microbial communities from our environ-
have been extensively studied, and although knowledge ment (particularly those of our microbiota) may help to
is rapidly expanding, the complexity of these interactions identify new bacterial species with beneficial effects on
is also becoming increasingly apparent. This has pre- human health. Interestingly, the recent description of
vented clear conclusions from being made about the ap- the intestinal metagenome (i.e. all genomes of the bac-
propriate markers to examine the effects of a food or a terial populations present in the intestine), by sequen-
microorganism on human health. As a result, a gap ex- cing strategies has confirmed that the microbial
ists between productive research and the effective imple- richness of the human gut microbiome correlates with
mentation of these findings in the life of the consumer. metabolic markers [75]. Therefore, besides being a
This is a good illustration of the challenges of transla- huge reservoir of unexploited commensal bacteria, our
tional research. In addition to research on probiotics microbiota also has metabolic capacities that are poten-
and their effects, there is also a strong and sustainable tially beneficial to human health. We can speculate that
need to document markers and to assess the clinical sig- this large panel of commensal organisms contains new
nificance of probiotics on human health. Hopefully, pro- promising candidates that may be very efficient in the
biotics have many potential activities and although digestive tract because they will be reintroduced into
efforts should focus on the microbiota and immune sys- their endogenous ecological niche. However, although
tem, other effects that are clearly beneficial to humans, the GI tract is their natural niche, most potentially
can be more readily assessed. A non-exhaustive list of commensal probiotic bacteria have never been used in
potential benefits includes reduction in cholesterol food products; thus, manufacturers would have to apply
levels, decrease of intestinal pain, favored intestinal tran- for marketing authorization according to the regulation
sit, and reinforcement of the intestinal epithelium. of novel foods.
Miquel et al. Microbial Cell Factories (2015) 14:48 Page 9 of 11

Conclusion 2. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al. Expert
We propose a framework that will help academic and in- consensus document: the international scientific association for probiotics
and prebiotics consensus statement on the scope and appropriate use of
dustrial communities to explore the potential of bacteria the term probiotic. Nat Rev Gastroenterol Hepatol. 2014;11(8):506–14.
as novel foods with health claims in accordance with 3. Schmidt C. The startup bugs. Nat Biotechnol. 2013;31(4):279–81.
European health and nutrition policy. New research on 4. Hungin AP, Mulligan C, Pot B, Whorwell P, Agreus L, Fracasso P, et al.
Systematic review: probiotics in the management of lower gastrointestinal
the human microbiome will facilitate the development symptoms in clinical practice - an evidence-based international guide.
of mechanistically-driven probiotics. This approach Aliment Pharmacol Ther. 2013;38(8):864–86.
clearly offers a new strategy that may benefit the health 5. Ritchie ML, Romanuk TN. A meta-analysis of probiotic efficacy for
gastrointestinal diseases. PLoS One. 2012;7(4):e34938.
of the general population and that of patients with lim- 6. Smug LN, Salminen S, Sanders ME, Ebner S. Yoghurt and probiotic bacteria
ited therapeutic options; for example, it may provide an in dietary guidelines of the member states of the European Union. Benef
alternative to long-term antibiotic use. Further insight Microbes. 2014;5(1):61–6.
7. Sanders ME, Lenoir-Wijnkoop I, Salminen S, Merenstein DJ, Gibson GR,
into the precise mechanisms of action of new probiotic Petschow BW, et al. Probiotics and prebiotics: prospects for public health
strains may lead to the development of second gener- and nutritional recommendations. Ann N Y Acad Sci. 2014;1309:19–29.
ation probiotics with characterized beneficial effects. 8. Foligne B, Daniel C, Pot B. Probiotics from research to market: the
possibilities, risks and challenges. Curr Opin Microbiol. 2013;16(3):284–92.
Until now, yoghurt is the only probiotic food with a 9. Bron PA, Tomita S, Mercenier A, Kleerebezem M. Cell surface-associated
health claim [11]. It remains unclear whether these regu- compounds of probiotic lactobacilli sustain the strain-specificity dogma.
lations limit or boost creativity and innovation [76]. It is Curr Opin Microbiol. 2013;16(3):262–9.
10. Turpin W, Humblot C, Thomas M, Guyot JP. Lactobacilli as multifaceted
in the interest of stakeholders that this translational sub-
probiotics with poorly disclosed molecular mechanisms. Int J Food
ject, at the cross roads of scientific, industrial, and clin- Microbiol. 2010;143(3):87–102.
ical research, is clarified by appropriate regulations [77]. 11. EFSA Panel on Dietetic Products, Nutrition and Allergies NDA. Scientific
These regulations clearly indicate, to companies as well Opinion on the substantiation of health claims related to live yoghurt
cultures and improved lactose digestion (ID 1143, 2976) pursuant to Article
as to risk assessors and managers, that claims should be 13(1) of Regulation (EC) No 1924/2006. EFSA Journal. 2010;8(10):1763.
based only on very strong scientific evidence. Moreover, 12. EFSA Panel on Dietetic Products, Nutrition and Allergies NDA. Scientific
it seems necessary to have some flexibility regarding in- Opinion on the substantiation of health claims related to non-characterised
bacteria and yeasts pursuant to Article 13(1) of Regulation (EC) No 1924/2006.
dividual studies depending on the particular microorgan- EFSA Journal. 2010;8(2):1470.
ism involved, the claim area, the target population, and 13. FAO/WHO. Guidelines for the Evaluation of Probiotics in Food. 2002.
the condition of use. In this “point of view” paper, we 14. Donelli G, Vuotto C, Mastromarino P. Phenotyping and genotyping are both
essential to identify and classify a probiotic microorganism. Microb Ecol
have discussed some of the tests proposed for the devel- Health Dis. 2013;24:20105 - http://dx.doi.org/10.3402/mehd.v24i0.20105.
opment of intestinal probiotics, bearing in mind that in- 15. Rijkers GT, de Vos WM, Brummer RJ, Morelli L, Corthier G, Marteau P. Health
novative strategies should be encouraged. benefits and health claims of probiotics: bridging science and marketing.
Br J Nutr. 2011;106(9):1291–6.
Competing interests 16. Kleerebezem M, Vaughan EE. Probiotic and gut lactobacilli and
The author(s) declare that they have no competing interests. bifidobacteria: molecular approaches to study diversity and activity. Annu
Rev Microbiol. 2009;63:269–90.
Authors’ contributions 17. Ben-Yahia L, Mayeur C, Rul F, Thomas M. Growth advantage of
SM, VB, MT raised the necessity to review Novel Food regulation and Streptococcus thermophilus over Lactobacillus bulgaricus in vitro and in the
conceived of the work. SM, MB, RM, MT drafted the manuscript, illustrations gastrointestinal tract of gnotobiotic rats. Benef Microbes. 2012;3(3):211–9.
and tables. MT coordinated the work, PL funded financial supports for SM 18. Delorme C. Safety assessment of dairy microorganisms: Streptococcus
and RM. PL, VB and MT provided the final version. All authors read and thermophilus. Int J Food Microbiol. 2008;126(3):274–7.
approved the final manuscript. 19. Herve-Jimenez L, Guillouard I, Guedon E, Gautier C, Boudebbouze S, Hols P,
et al. Physiology of Streptococcus thermophilus during the late stage of
Acknowledgements milk fermentation with special regard to sulfur amino-acid metabolism.
We thank Claire Cherbuy, Muriel Mercier-Bonin, Françoise Rul, and Joanna Proteomics. 2008;8(20):4273–86.
Radziwiłł-Bieńkowska for fruitful discussions. SM and RM received funding 20. Pineiro M, Stanton C. Probiotic bacteria: legislative framework–
from the FPARIS collaborative project, which was selected and supported by requirements to evidence basis. J Nutr. 2007;137(3 Suppl 2):850S–3.
the Vitagora Competitive Cluster and funded by the French FUI (Fond 21. Sanders ME, Akkermans LM, Haller D, Hammerman C, Heimbach J,
Unique Interministériel; FUI: n°F1010012D), the FEDER (Fonds Européen de Hormannsperger G, et al. Safety assessment of probiotics for human use.
Développement Régional; Bourgogne: 34606), the Burgundy Region, the Gut Microbes. 2010;1(3):164–85.
Conseil Général 21, and the Grand Dijon. 22. Guarner F, Khan AG, Garisch J, Eliakim R, Gangl A, Thomson A, et al. World
Gastroenterology Organisation Global Guidelines: probiotics and prebiotics
Author details October 2011. J Clin Gastroenterol. 2012;46(6):468–81.
1
Commensal and Probiotics-Host Interactions Laboratory, UMR1319 Micalis, 23. EFSA Panel on Dietetic Products, Nutrition and Allergies (NDA). General
INRA, AgroParisTech, Domaine de Vilvert, 78350 Jouy en Josas, France. guidance for stakeholders on the evaluation of Article 13.1, 13.5 and 14
2
AgroParisTech, UMR 1319 MICALIS, F-78350 Jouy-en-Josas, France. health claims. EFSA Journal. 2011;9(4):2135.
3
VAB-nutrition, Clermont-Ferrand, France. 24. Mitropoulou G, Nedovic V, Goyal A, Kourkoutas Y. Immobilization
technologies in probiotic food production. J Nutr Metab. 2013;2013:716861.
Received: 14 November 2014 Accepted: 17 March 2015 25. Rousseaux C, Thuru X, Gelot A, Barnich N, Neut C, Dubuquoy L, et al.
Lactobacillus acidophilus modulates intestinal pain and induces opioid and
cannabinoid receptors. Nat Med. 2007;13(1):35–7.
References 26. EFSA Panel on Biological Hazards. Scientific Opinion on the maintenance of
1. FAO/WHO. Health and nutritional properties of probiotics in food including the list of QPS biological agents intentionally added to food and feed
powder milk with live lactic acid bacteria. 2001. (2013 update). EFSA Journal. 2013;11:3449.
Miquel et al. Microbial Cell Factories (2015) 14:48 Page 10 of 11

27. Adams MR, Marteau P. On the safety of lactic acid bacteria from food. Int J 52. Duncan SH, Louis P, Flint HJ. Lactate-utilizing bacteria, isolated from human
Food Microbiol. 1995;27(2–3):263–4. feces, that produce butyrate as a major fermentation product. Appl Environ
28. Joly F, Mayeur C, Bruneau A, Noordine ML, Meylheuc T, Langella P, et al. Microbiol. 2004;70(10):5810–7.
Drastic changes in fecal and mucosa-associated microbiota in adult patients 53. de Vrese M, Barth CA. Postprandial plasma D-lactate concentrations after
with short bowel syndrome. Biochimie. 2010;92(7):753–61. yogurt ingestion. Z Ernahrungswiss. 1991;30(2):131–7.
29. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. A human 54. European Commission Health and Consumer Protection Directorate-
gut microbial gene catalogue established by metagenomic sequencing. General, Scientific Committee on Animal Nutrition (SCAN). Opinion of the
Nature. 2010;464(7285):59–65. scientific committee on animal nutrition on the safety of use of Bacillus
30. Marteau P. Safety aspects of probiotic products. Scand J Nutr. 2001;45:22–4. species in animal nutrition. 2000.
31. Salminen S, von Wright A, Morelli L, Marteau P, Brassart D, de Vos WM, et al. 55. Al Kassaa I, Hamze M, Hober D, Chihib NE, Drider D. Identification of vaginal
Demonstration of safety of probiotics – a review. Int J Food Microbiol. lactobacilli with potential probiotic properties isolated from women in
1998;44(1–2):93–106. North Lebanon. Microb Ecol. 2014;67(3):722–34.
32. Silley P. Do bacteria need to be regulated? J Appl Microbiol. 2006;101(3):607–15. 56. Del Turco S, Sartini S, Cigni G, Sentieri C, Sbrana S, Battaglia D, et al.
33. Borchers AT, Selmi C, Meyers FJ, Keen CL, Gershwin ME. Probiotics and Synthetic analogues of flavonoids with improved activity against platelet
immunity. J Gastroenterol. 2009;44(1):26–46. activation and aggregation as novel prototypes of food supplements. Food
34. Mater DD, Bretigny L, Firmesse O, Flores MJ, Mogenet A, Bresson JL, et al. Chem. 2015;175:494–9.
Streptococcus thermophilus and Lactobacillus delbrueckii subsp. bulgaricus 57. Suslova TE, Sitozhevskii AV, Ogurkova ON, Kravchenko ES, Kologrivova IV,
survive gastrointestinal transit of healthy volunteers consuming yogurt. Anfinogenova Y, et al. Platelet hemostasis in patients with metabolic
FEMS Microbiol Lett. 2005;250(2):185–7. syndrome and type 2 diabetes mellitus: cGMP- and NO-dependent mechanisms
35. Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis factor prevents in the insulin-mediated platelet aggregation. Front Physiol. 2015;5:501.
intestinal inflammatory disease. Nature. 2008;453(7195):620–5. 58. Bangemann M. 97/618/CE: Recommandation de la Commission du 29 juillet
36. Macho Fernandez E, Valenti V, Rockel C, Hermann C, Pot B, Boneca IG, et al. 1997. Journal officiel. 1997;L 253:1–36.
Anti-inflammatory capacity of selected lactobacilli in experimental colitis is 59. de Graaf IA, Olinga P, de Jager MH, Merema MT, de Kanter R, van de
driven by NOD2-mediated recognition of a specific peptidoglycan-derived Kerkhof EG, et al. Preparation and incubation of precision-cut liver and
muropeptide. Gut. 2011;60(8):1050–9. intestinal slices for application in drug metabolism and toxicity studies. Nat
37. Riaz QU, Masud T. Recent trends and applications of encapsulating Protoc. 2010;5(9):1540–51.
materials for probiotic stability. Crit Rev Food Sci Nutr. 2013;53(3):231–44. 60. Leushacke M, Barker N. Ex vivo culture of the intestinal epithelium:
38. Vyas U, Ranganathan N. Probiotics, prebiotics, and synbiotics: gut and strategies and applications. Gut. 2014;63(8):1345–54.
beyond. Gastroenterol Res Pract. 2012;2012:872716. 61. EFSA Panel on Dietetic Products, Nutrition and Allergies (NDA). Guidance on
39. Grill JP, Perrin S, Schneider F. Bile salt toxicity to some bifidobacteria strains: the scientific requirements for health claims related to gut and immune
role of conjugated bile salt hydrolase and pH. Can J Microbiol. 2000;46 function. EFSA Journal. 2011;9(4):12.
(10):878–84. 62. EFSA Panel on Dietetic Products, Nutrition and Allergies (NDA). Guidance on
40. Begley M, Hill C, Gahan CG. Bile salt hydrolase activity in probiotics. Appl the scientific requirements for health claims related to antioxidants,
Environ Microbiol. 2006;72(3):1729–38. oxidative damage and cardiovascular health. EFSA Journal. 2011;9(12):13.
41. EFSA Panel on Additives and Products, or Substances used in Animal Feed 63. EFSA Panel on Dietetic Products, Nutrition and Allergies (NDA). Guidance on
FEEDAP. Guidance on the assessment of bacterial susceptibility to the scientific requirements for health claims related to appetite ratings, weight
antimicrobials of human and veterinary importance. EFSA Journal. management, and blood glucose concentrations. EFSA Journal. 2011;10(3):11.
2012;10(6):2740. 64. The Human Microbiome Project Consortium. Structure, function and
42. Bayliss CE, Houston AP. Characterization of plant polysaccharide- and diversity of the healthy human microbiome. Nature. 2012;486(7402):207–14.
mucin-fermenting anaerobic bacteria from human feces. Appl Environ 65. Aziz Q, Dore J, Emmanuel A, Guarner F, Quigley EM. Gut microbiota and
Microbiol. 1984;48(3):626–32. gastrointestinal health: current concepts and future directions.
43. Wrzosek L, Miquel S, Noordine ML, Bouet S, Chevalier-Curt MJ, Robert V, Neurogastroenterol Motil. 2013;25(1):4–15.
et al. Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii 66. Cherbuy C, Honvo-Houeto E, Bruneau A, Bridonneau C, Mayeur C, Duee PH,
influence the production of mucus glycans and the development of goblet et al. Microbiota matures colonic epithelium through a coordinated
cells in the colonic epithelium of a gnotobiotic model rodent. BMC Biol. induction of cell cycle-related proteins in gnotobiotic rat. Am J Physiol
2013;11:61. Gastrointest Liver Physiol. 2010;299(2):G348–57.
44. Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, et al. Cross- 67. Gaboriau-Routhiau V, Rakotobe S, Lecuyer E, Mulder I, Lan A, Bridonneau C,
talk between Akkermansia muciniphila and intestinal epithelium controls et al. The key role of segmented filamentous bacteria in the coordinated
diet-induced obesity. Proc Natl Acad Sci U S A. 2013;110(22):9066–71. maturation of gut helper T cell responses. Immunity. 2009;31(4):677–89.
45. Guidance for Industry. Early Clinical Trials with Live Biotherapeutic Products: 68. Tomas J, Reygner J, Mayeur C, Ducroc R, Bouet S, Bridonneau C, et al. Early
Chemistry, Manufacturing, and Control Information. http://wwwfdagov/ colonizing Escherichia coli elicits remodeling of rat colonic epithelium
downloads/BiologicsBloodVaccines/Guidance-ComplianceRegulatoryInformation/ shifting toward a new homeostatic state. ISME J. 2015;9(1):46–58.
Guidances/General/UCM292704pdf February 2012. 69. Tomas J, Wrzosek L, Bouznad N, Bouet S, Mayeur C, Noordine ML, et al.
46. McNulty NP, Yatsunenko T, Hsiao A, Faith JJ, Muegge BD, Goodman AL, Primocolonization is associated with colonic epithelial maturation during
et al. The impact of a consortium of fermented milk strains on the gut conventionalization. Faseb J. 2013;27(2):645–55.
microbiome of gnotobiotic mice and monozygotic twins. Sci Transl Med. 70. De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S,
2011;3(106):106ra106. et al. Impact of diet in shaping gut microbiota revealed by a comparative
47. Hapfelmeier S, Lawson MA, Slack E, Kirundi JK, Stoel M, Heikenwalder M, study in children from Europe and rural Africa. Proc Natl Acad Sci U S A.
et al. Reversible microbial colonization of germ-free mice reveals the 2010;107(33):14691–6.
dynamics of IgA immune responses. Science. 2010;328(5986):1705–9. 71. Miquel S, Martin R, Bridonneau C, Robert V, Sokol H, Bermudez-Humaran LG,
48. Turpin W, Humblot C, Noordine ML, Thomas M, Guyot JP. Lactobacillaceae et al. Ecology and metabolism of the beneficial intestinal commensal
and cell adhesion: genomic and functional screening. PLoS One. bacterium. Gut Microbes. 2014;5(2):146–51.
2012;7(5):e38034. 72. Miquel S, Martin R, Rossi O, Bermudez-Humaran LG, Chatel JM, Sokol H,
49. Ledoux D, Labombardi VJ, Karter D. Lactobacillus acidophilus bacteraemia et al. Faecalibacterium prausnitzii and human intestinal health. Curr Opin
after use of a probiotic in a patient with AIDS and Hodgkin’s disease. Int J Microbiol. 2013;16(3):255–61.
STD AIDS. 2006;17(4):280–2. 73. Martin R, Chain F, Miquel S, Lu J, Gratadoux JJ, Sokol H, et al. The
50. Mack DR. D(−)-lactic acid-producing probiotics, D(−)-lactic acidosis and commensal bacterium Faecalibacterium prausnitzii is protective in DNBS-
infants. Can J Gastroenterol. 2004;18(11):671–5. induced chronic moderate and severe colitis models. Inflamm Bowel Dis.
51. Mayeur C, Gratadoux JJ, Bridonneau C, Chegdani F, Larroque B, Kapel N, et al. 2014;20(3):417–30.
Faecal D/L lactate ratio is a metabolic signature of microbiota imbalance in 74. Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermudez-Humaran LG,
patients with short bowel syndrome. PLoS One. 2013;8(1):e54335. Gratadoux JJ, et al. Faecalibacterium prausnitzii is an anti-inflammatory
Miquel et al. Microbial Cell Factories (2015) 14:48 Page 11 of 11

commensal bacterium identified by gut microbiota analysis of Crohn


disease patients. Proc Natl Acad Sci U S A. 2008;105(43):16731–6.
75. Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, et al.
Richness of human gut microbiome correlates with metabolic markers.
Nature. 2013;500(7464):541–6.
76. van Loveren H, Sanz Y, Salminen S. Health claims in Europe: probiotics and
prebiotics as case examples. Annu Rev Food Sci Technol. 2012;3:247–61.
77. Arnold C. The pros and cons of probiotics. Lancet Infect Dis. 2013;13(7):571–2.

Submit your next manuscript to BioMed Central


and take full advantage of:

• Convenient online submission


• Thorough peer review
• No space constraints or color figure charges
• Immediate publication on acceptance
• Inclusion in PubMed, CAS, Scopus and Google Scholar
• Research which is freely available for redistribution

Submit your manuscript at


www.biomedcentral.com/submit

You might also like